ALA7 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of ALA7 Antibody

The term "ALA7 Antibody" refers to monoclonal antibodies targeting Activin Receptor-Like Kinase 7 (ALK7), a type I receptor in the TGF-β superfamily. ALK7 is highly expressed in adipose tissue and plays critical roles in lipid metabolism, insulin regulation, and obesity-related pathways . Neutralizing ALK7 antibodies, such as the fully human IgG1 monoclonal antibody described in preclinical studies, have demonstrated therapeutic potential for metabolic disorders by enhancing lipolysis and reducing adiposity .

Key Effects in Obesity Models:

ParameterALK7 Antibody vs. ControlSource
Body weight reduction50% decrease in adiposity
Fat pad weight40–60% reduction
Serum leptin levelsProportional decrease
Glucose intoleranceMarked improvement
  • In diet-induced obese mice, ALK7 antibody treatment increased whole-body oxygen consumption and fatty acid oxidation in muscle .

  • The antibody reduced NLRP3 inflammasome activation in adipose tissue macrophages (ATMs), lowering systemic inflammation .

Clinical Relevance and Applications

Metabolic Disorders:

  • ALK7 inhibition enhances lipolysis, counteracting obesity and insulin resistance .

  • Human genetic studies link ALK7 variants to reduced diabetes risk, validating its therapeutic potential .

Autoimmune/Inflammatory Conditions:

  • Antilymphocyte antibodies (ALA) targeting T cells have been reported in scleroderma and HIV, though their relationship to ALK7-specific antibodies requires further study .

Challenges and Future Directions

  • Safety: No cytotoxic effects (e.g., elevated LDH) were observed in murine models .

  • Delivery Optimization: Antibody distribution remains adipose-tissue-preferential, necessitating targeted delivery systems for broader applications .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
ALA7 antibody; At3g13900 antibody; MDC16.2 antibody; Probable phospholipid-transporting ATPase 7 antibody; AtALA7 antibody; EC 7.6.2.1 antibody; Aminophospholipid flippase 7 antibody
Target Names
ALA7
Uniprot No.

Target Background

Function
This antibody targets a protein involved in phospholipid transport and the regulation of pollen plasma membrane lipid asymmetry.
Database Links
Protein Families
Cation transport ATPase (P-type) (TC 3.A.3) family, Type IV subfamily
Subcellular Location
Cell membrane; Multi-pass membrane protein. Endomembrane system; Multi-pass membrane protein.

Q&A

What is [D-Ala⁷]-Angiotensin I/II (1-7) and what role do antibodies against it play in research?

[D-Ala⁷]-Angiotensin I/II (1-7) is a synthetic peptide that functions as a potent and selective Angiotensin (1-7) Mas receptor antagonist. It has the ability to antagonize several actions of Ang-(1-7), including the antidiuretic effect of the peptide and the changes in mean arterial pressure produced by Ang-(1-7) microinjection into specific brain regions . Antibodies against this compound are valuable research tools that allow scientists to detect, quantify, and study the distribution and functions of [D-Ala⁷]-Angiotensin I/II (1-7) in biological systems. These antibodies enable researchers to investigate the mechanisms by which [D-Ala⁷]-Angiotensin I/II (1-7) modulates the renin-angiotensin system (RAS), which plays crucial roles in cardiovascular regulation, fluid balance, and blood pressure control .

How do antibodies against [D-Ala⁷]-Angiotensin I/II (1-7) differ from other angiotensin receptor antibodies?

Antibodies against [D-Ala⁷]-Angiotensin I/II (1-7) are specifically designed to recognize this synthetic peptide antagonist, which targets the Angiotensin-(1-7) Mas receptor. These antibodies differ from antibodies against other angiotensin receptors (such as AT1 or AT2 receptors) in their epitope recognition, binding specificity, and research applications . While AT1 receptor antibodies target the classical pathway of RAS that promotes vasoconstriction and sodium retention, antibodies against [D-Ala⁷]-Angiotensin I/II (1-7) are valuable for studying the counter-regulatory axis of RAS involving Ang-(1-7) . Researchers must carefully validate the specificity of these antibodies to ensure they don't cross-react with other components of the RAS, as this system contains several peptides and receptors with structural similarities .

What are the key experimental applications of antibodies targeting [D-Ala⁷]-Angiotensin I/II (1-7)?

Antibodies targeting [D-Ala⁷]-Angiotensin I/II (1-7) have several important experimental applications in research settings. These include:

  • Immunohistochemistry and immunofluorescence to visualize the distribution of [D-Ala⁷]-Angiotensin I/II (1-7) in tissue sections

  • Western blotting for detecting and quantifying the peptide in protein samples

  • Immunoprecipitation to isolate [D-Ala⁷]-Angiotensin I/II (1-7) and its binding partners

  • ELISA for quantitative measurement in biological fluids

  • Flow cytometry to analyze cellular expression patterns

These antibodies are particularly valuable in studying the effects of [D-Ala⁷]-Angiotensin I/II (1-7) on blood pressure regulation, renal function, and cardiovascular health, as the peptide antagonizes the antidiuretic effect of Ang-(1-7) and affects mean arterial pressure . Researchers often use these antibodies alongside functional studies to correlate the presence of the peptide with its physiological effects.

What are the recommended methods for optimizing antibody specificity against [D-Ala⁷]-Angiotensin I/II (1-7)?

Optimizing antibody specificity against [D-Ala⁷]-Angiotensin I/II (1-7) requires a multi-faceted approach. Researchers should consider implementing machine learning and deep sequencing techniques to identify and select antibody variants with optimal binding properties . An integrated experimental and computational approach that combines deep sequencing with machine learning can help predict antibody mutations at novel CDR sites that co-optimize multiple properties linked to therapeutic antibody performance . Linear discriminant analysis (LDA) models trained with deep learning features have shown superior performance at generalizing to novel mutational space compared to conventional physicochemical antibody features .

To validate specificity, researchers should perform competitive binding assays with structurally similar peptides (like Ang II and native Ang-(1-7)) to ensure the antibody preferentially recognizes [D-Ala⁷]-Angiotensin I/II (1-7) . Cross-reactivity testing against related peptides in the RAS is essential, and techniques such as phage-display technology can be employed to generate highly specific antibodies, similar to approaches used for other target proteins .

How can researchers best validate the functionality of antibodies against [D-Ala⁷]-Angiotensin compounds in cellular and animal models?

Validation of antibodies against [D-Ala⁷]-Angiotensin compounds requires comprehensive testing in both cellular and animal models. In cellular systems, researchers should:

  • Verify binding specificity using cells expressing the Mas receptor versus control cells

  • Assess the antibody's ability to detect changes in [D-Ala⁷]-Angiotensin levels after relevant stimuli

  • Confirm the antibody's capacity to block the antagonistic effects of [D-Ala⁷]-Angiotensin on Ang-(1-7) signaling

In animal models, particularly those relevant to hypertension or renal disease, validation should include:

  • Immunohistochemical analysis to confirm appropriate tissue distribution

  • Physiological readouts to correlate antibody binding with functional effects, such as changes in blood pressure or renal function

  • Dose-response studies using the antibody in animals treated with [D-Ala⁷]-Angiotensin I/II (1-7) (typically administered at 5 mg/kg intraperitoneally in mice)

  • Comparative studies in wildtype versus Mas receptor knockout animals to confirm specificity

Functional validation can be performed by testing whether the antibody blocks the antagonistic effects of [D-Ala⁷]-Angiotensin I/II (1-7) on Ang-(1-7)-mediated responses in relevant physiological systems .

What techniques are most effective for quantitative analysis of [D-Ala⁷]-Angiotensin I/II (1-7) using antibody-based methods?

For quantitative analysis of [D-Ala⁷]-Angiotensin I/II (1-7), several antibody-based techniques have proven effective. Enzyme-linked immunosorbent assays (ELISA) offer high sensitivity and specificity for quantifying the peptide in biological fluids or tissue extracts. Researchers should develop calibration curves using purified [D-Ala⁷]-Angiotensin I/II (1-7) standards and validate the assay's linearity, precision, and accuracy .

Radioimmunoassays using radiolabeled [D-Ala⁷]-Angiotensin I/II (1-7) can provide excellent sensitivity for detecting low concentrations in complex samples. Additionally, advanced mass spectrometry techniques coupled with immunoprecipitation using anti-[D-Ala⁷]-Angiotensin antibodies can provide highly specific quantification .

For in situ quantification, quantitative immunofluorescence using calibrated imaging systems allows researchers to measure relative concentrations of the peptide in tissue sections while preserving spatial information . When analyzing receptor binding, techniques similar to those used for other receptor-ligand interactions can be adapted, such as monitoring calcium responses through elevation in aequorin-derived fluorescence in response to increasing concentrations of [D-Ala⁷]-Angiotensin I/II (1-7) .

How can antibodies against [D-Ala⁷]-Angiotensin compounds be utilized to study the counterregulatory axis of the renin-angiotensin system?

Antibodies against [D-Ala⁷]-Angiotensin compounds provide powerful tools for investigating the counterregulatory axis of the renin-angiotensin system. By selectively binding to these antagonists, researchers can manipulate and study the balance between the classical RAS pathway (mediated by Ang II and AT1 receptors) and the counterregulatory pathway (mediated by Ang-(1-7) and Mas receptors) . These antibodies enable researchers to neutralize [D-Ala⁷]-Angiotensin I/II (1-7) in experimental settings, effectively removing its antagonistic effects on Ang-(1-7), which allows for the investigation of the physiological roles of endogenous Ang-(1-7) .

In renal research, these antibodies have been instrumental in elucidating how Ang-(1-7) counteracts the effects of Ang II on sodium reabsorption, vasoconstriction, and fibrotic processes . Studies have shown that blockade with the Ang-(1-7) antagonist [D-Ala⁷]-Ang-(1-7) affects renal function and blood pressure regulation, particularly in animal models maintained on salt-restricted diets . By using these antibodies in combination with molecular and physiological techniques, researchers can dissect the complex interplay between different RAS components and their impact on cardiovascular and renal health .

What are the challenges and solutions in developing therapeutic antibodies targeting the [D-Ala⁷]-Angiotensin pathway?

Developing therapeutic antibodies targeting the [D-Ala⁷]-Angiotensin pathway presents several challenges. The small size of the peptide makes it difficult to generate highly specific antibodies with therapeutic potential. Additionally, the structural similarity between [D-Ala⁷]-Angiotensin I/II (1-7) and other angiotensin peptides increases the risk of cross-reactivity and off-target effects .

To address these challenges, researchers can employ advanced antibody engineering approaches. The integration of experimental and computational methods that combine deep sequencing, machine learning, and high-throughput techniques has shown promise in optimizing both antibody affinity and specificity . Linear discriminant analysis (LDA) models trained with deep learning features have demonstrated superior performance in predicting beneficial antibody mutations compared to conventional approaches .

Another challenge is maintaining the delicate balance of the RAS, as complete blockade of [D-Ala⁷]-Angiotensin effects might disrupt normal physiological processes. Developing antibodies with tunable binding properties or context-dependent activity could help address this issue . For therapeutic applications, fully human antibodies similar to those developed for other targets (like the anti-IL-7Rα antibody) could minimize immunogenicity concerns .

How can multi-objective optimization techniques improve the development of antibodies against [D-Ala⁷]-Angiotensin compounds?

Multi-objective optimization techniques offer significant advantages for developing improved antibodies against [D-Ala⁷]-Angiotensin compounds. These approaches simultaneously optimize multiple antibody properties, such as affinity, specificity, stability, and manufacturability, rather than focusing on a single attribute . Linear discriminant analysis (LDA) models can generate one-dimensional projections that reflect the continuous variability of multiple biophysical properties, enabling the direct identification of Pareto optimal antibodies that represent the best possible trade-offs between competing properties .

This approach is particularly valuable because there are often strict tradeoffs between antibody properties (such as affinity and specificity) at the library scale. By implementing computational models trained on binary datasets for affinity and specificity, researchers can predict continuous property values that facilitate multi-objective optimization . For [D-Ala⁷]-Angiotensin antibodies, this could mean developing variants with optimal combinations of target binding affinity, reduced cross-reactivity with other angiotensin peptides, and suitable pharmacokinetic properties.

The integration of deep learning features in LDA models has proven superior at generalizing to novel mutational space compared to conventional physicochemical antibody features, making this approach promising for engineering next-generation antibodies against [D-Ala⁷]-Angiotensin compounds .

What are the optimal experimental conditions for using antibodies against [D-Ala⁷]-Angiotensin in various research techniques?

When using antibodies against [D-Ala⁷]-Angiotensin in research, optimal experimental conditions vary by technique but follow several general principles. For immunohistochemistry and immunofluorescence, researchers should optimize fixation methods (typically 4% paraformaldehyde), antigen retrieval (if needed), blocking solutions (5-10% normal serum), antibody concentration (typically 1-10 μg/ml), and incubation conditions (4°C overnight) . Primary antibody specificity should be validated using appropriate positive and negative controls, including tissues known to express or lack the target .

For Western blotting, optimal conditions include effective protein extraction methods (using protease inhibitors to prevent degradation of the peptide), appropriate blocking buffers (typically 5% BSA), antibody dilution (determined through titration experiments), and incubation times . For ELISA, researchers should optimize coating conditions, blocking reagents, antibody concentrations, and detection systems to achieve maximum sensitivity and specificity .

When studying receptor-ligand interactions, conditions similar to those used in calcium response assays for AT1R activation can be employed, where researchers monitor elevation in aequorin-derived fluorescence in response to increasing concentrations of [D-Ala⁷]-Angiotensin I/II (1-7) . All experiments should include appropriate controls to account for non-specific binding and background signal.

How can researchers troubleshoot common issues with antibodies against [D-Ala⁷]-Angiotensin compounds?

Researchers working with antibodies against [D-Ala⁷]-Angiotensin compounds may encounter several common issues. High background signal can be addressed by optimizing blocking conditions (trying different blocking agents like BSA, milk, or normal serum), increasing washing steps, and titrating the antibody concentration to find the optimal signal-to-noise ratio . Cross-reactivity with other angiotensin peptides can be minimized by pre-absorbing the antibody with potential cross-reactive antigens or by using more specific monoclonal antibodies .

For weak or no signal, researchers should verify that their sample preparation methods preserve the antigen, consider alternative antigen retrieval methods for fixed tissues, and test different antibody clones or detection systems . If antibody recognition is inconsistent across different experimental platforms, it may indicate conformation-dependent epitope recognition, which can be addressed by using denaturing or native conditions as appropriate .

For functional studies, if the antibody fails to block the expected [D-Ala⁷]-Angiotensin effects, researchers should verify that the antibody is being used at sufficient concentration and that the experimental readout is appropriate for detecting the antagonistic effects of [D-Ala⁷]-Angiotensin I/II (1-7) on Ang-(1-7) signaling .

What control experiments are essential when using antibodies against [D-Ala⁷]-Angiotensin in research?

When using antibodies against [D-Ala⁷]-Angiotensin in research, several essential control experiments should be performed to ensure result validity. Specificity controls include competitive inhibition with excess purified [D-Ala⁷]-Angiotensin I/II (1-7) peptide, which should abolish specific antibody binding, and testing the antibody against samples known to contain or lack the target (positive and negative controls) .

Isotype controls using non-specific antibodies of the same isotype as the primary antibody are crucial to distinguish specific binding from Fc receptor interactions or other non-specific binding . For functional studies, comparing the effects of the anti-[D-Ala⁷]-Angiotensin antibody with other established antagonists of the Ang-(1-7)/Mas receptor pathway can provide validation of the antibody's functional effects .

When studying in vivo effects, controls should include experiments in genetic models where components of the RAS system are knocked out or modified, such as comparing wildtype mice to mas knockout mice . Dose-response experiments are also essential to establish the optimal antibody concentration for specific applications and to distinguish specific from non-specific effects . Finally, when using multiple detection methods, researchers should verify that results are consistent across different experimental platforms to rule out technique-specific artifacts.

How should researchers interpret contradictory findings when using antibodies against [D-Ala⁷]-Angiotensin in different experimental systems?

When researchers encounter contradictory findings using antibodies against [D-Ala⁷]-Angiotensin across different experimental systems, careful analysis is required. First, examine the specific experimental conditions in each system, as the RAS exhibits context-dependent regulation. The effects of [D-Ala⁷]-Angiotensin I/II (1-7) can vary depending on the physiological state of the animal model, particularly in studies focused on renal function and fluid balance . Multiple renal actions on fluid balance have been shown for the AT(1-7) receptor, and these actions vary depending on the physiological state of the animal .

Consider the antibody clone, concentration, and detection method used in each study, as these factors can significantly impact results. Different epitopes may be accessible depending on sample preparation, leading to apparent contradictions . The specificity and cross-reactivity profile of the antibody should be thoroughly assessed in each experimental system, particularly when working with structurally similar peptides in the RAS .

Additionally, evaluate the cellular context, as the signaling pathways and effects of Ang-(1-7) can differ between cell types. For example, in proximal tubule cells, Ang-(1-7) reduces Ang II-mediated phosphorylation of proteins such as p38, ERK 1/2, and JNK, but these effects may differ in other cell types . When contradictions persist despite controlling for these variables, consider designing experiments that directly compare the different systems under identical conditions to identify the source of the discrepancy.

What advanced analytical techniques can enhance the utility of antibodies against [D-Ala⁷]-Angiotensin in research?

Advanced analytical techniques can significantly enhance the utility of antibodies against [D-Ala⁷]-Angiotensin in research. Super-resolution microscopy techniques, such as STORM, PALM, or SIM, provide nanoscale visualization of [D-Ala⁷]-Angiotensin distribution and co-localization with receptors and signaling molecules, offering insights beyond conventional microscopy . Proximity ligation assays can detect protein-protein interactions involving [D-Ala⁷]-Angiotensin I/II (1-7) with high sensitivity and specificity, revealing its binding partners in situ .

Mass spectrometry immunoprecipitation techniques combine the specificity of antibody-based capture with the analytical power of mass spectrometry for comprehensive characterization of [D-Ala⁷]-Angiotensin and its modifications in biological samples . Multiplexed immunoassays allow simultaneous detection of multiple components of the RAS, providing a systems-level view of how [D-Ala⁷]-Angiotensin interacts with other elements of the pathway .

Computational approaches, such as machine learning and LDA models trained with deep learning features, can predict antibody mutations that optimize multiple properties simultaneously, leading to improved reagents for studying [D-Ala⁷]-Angiotensin compounds . These advanced techniques provide researchers with more comprehensive and precise tools for investigating the complex roles of [D-Ala⁷]-Angiotensin in physiological and pathological processes.

How can researchers correlate antibody binding data with functional effects of [D-Ala⁷]-Angiotensin compounds in complex physiological systems?

Correlating antibody binding data with the functional effects of [D-Ala⁷]-Angiotensin compounds in complex physiological systems requires a multi-modal approach. Researchers should combine quantitative antibody binding measurements (using techniques like ELISA, radioimmunoassay, or flow cytometry) with functional readouts specific to the system being studied . For cardiovascular studies, measurements of blood pressure, vascular tone, and cardiac function provide relevant functional endpoints to correlate with antibody binding .

In renal studies, researchers can correlate antibody binding data with measurements of glomerular filtration rate, sodium excretion, and markers of renal damage . The effect of [D-Ala⁷]-Angiotensin I/II (1-7) on antagonizing the antidiuretic effects of Ang-(1-7) provides a functional readout that can be correlated with antibody binding in the kidney .

At the cellular level, signaling pathway activation can be measured alongside antibody binding. For example, researchers can correlate antibody binding with the inhibition of Ang II-stimulated phosphorylation of proteins such as p38, ERK 1/2, and JNK in proximal tubule cells, or with the reduction in TGF-β production . Mathematical modeling can help integrate binding and functional data across different scales, from molecular interactions to whole-organism physiology, providing a more comprehensive understanding of how [D-Ala⁷]-Angiotensin compounds modulate complex physiological systems .

What are the emerging technologies that will shape future research using antibodies against [D-Ala⁷]-Angiotensin compounds?

Emerging technologies are poised to revolutionize research using antibodies against [D-Ala⁷]-Angiotensin compounds. Advanced computational approaches, including machine learning and deep sequencing, will enable more efficient development of highly specific antibodies with optimized properties for detecting and manipulating [D-Ala⁷]-Angiotensin compounds . Linear discriminant analysis (LDA) models trained with deep learning features have already demonstrated superior performance in predicting beneficial antibody mutations compared to conventional approaches .

Single-cell technologies will allow researchers to study the heterogeneity in responses to [D-Ala⁷]-Angiotensin compounds at unprecedented resolution, revealing cell type-specific effects that may be masked in bulk tissue analyses . Antibody engineering techniques, such as those used to develop antibody-drug conjugates, could be applied to create dual-function reagents that not only bind to [D-Ala⁷]-Angiotensin compounds but also deliver therapeutic cargoes to specific tissues .

CRISPR-based technologies might be combined with antibody approaches to modulate the expression of [D-Ala⁷]-Angiotensin-related genes while simultaneously monitoring peptide levels using antibody-based detection. Advances in structural biology, including cryo-EM and computational modeling, will provide deeper insights into the molecular interactions between antibodies and [D-Ala⁷]-Angiotensin compounds, facilitating the design of antibodies with enhanced specificity and affinity .

What are the key research questions that remain unanswered regarding antibodies against [D-Ala⁷]-Angiotensin compounds?

Despite significant progress, several key research questions remain unanswered regarding antibodies against [D-Ala⁷]-Angiotensin compounds. The exact epitopes recognized by these antibodies and how epitope specificity correlates with functional effects are not fully understood . Understanding these relationships could lead to the development of more precise tools for studying specific aspects of [D-Ala⁷]-Angiotensin biology.

The potential of antibodies against [D-Ala⁷]-Angiotensin as therapeutic agents for conditions involving dysregulation of the RAS, such as hypertension, heart failure, and kidney disease, remains largely unexplored . The tissue-specific distribution and actions of [D-Ala⁷]-Angiotensin compounds, particularly in pathological states, need further investigation using specialized antibodies that can detect the peptide in complex tissue environments .

The interactions between [D-Ala⁷]-Angiotensin and other components of the RAS system beyond the Mas receptor, such as potential cross-talk with AT1 and AT2 receptors, remain to be fully elucidated . Additionally, the potential for antibodies against [D-Ala⁷]-Angiotensin to serve as biomarkers or diagnostic tools for conditions associated with RAS dysregulation warrants further investigation .

How can researchers contribute to standardizing antibody use in [D-Ala⁷]-Angiotensin research to improve reproducibility?

Researchers can contribute to standardizing antibody use in [D-Ala⁷]-Angiotensin research through several key practices. Detailed reporting of antibody characteristics is essential, including catalog numbers, clone designations, lot numbers, validation data, and specific experimental conditions used . Researchers should perform and report comprehensive validation studies for each antibody, including specificity testing against related peptides, dose-response relationships, and functionality in relevant experimental systems .

The development of reference standards and protocols specific to [D-Ala⁷]-Angiotensin research would facilitate comparison across studies. This could include standardized positive and negative controls, consistent sample preparation methods, and agreed-upon experimental readouts . Researchers should engage in collaborative initiatives to validate antibodies across multiple laboratories, similar to multicenter validation studies performed for other important research antibodies .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.